• No results found

Cover Page The handle

N/A
N/A
Protected

Academic year: 2021

Share "Cover Page The handle"

Copied!
10
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

Cover Page

The handle

http://hdl.handle.net/1887/80759

holds various files of this Leiden University

dissertation.

Author: Grand Moursel, L.

Title: Molecular pathology of hereditary cerebral hemorrhage with amyloidosis-Dutch

type

(2)

Discussion

(3)

137 136

Discussion

Aim of the study & central findings

The general aim of this thesis was to decipher the molecular pathogenesis of HCHWA-D. Since no proven therapeutic treatment exists to prevent or even delay the disease onset, the understanding of underlying pathomechanisms in HCHWA-D is important. It may help discovering new therapeutic targets and biomarkers that can be used to assess the efficacy of candidate drugs in treatment trials.

The main finding of the work presented in this thesis is that TGFβ deregulation plays a central role in HCHWA-D pathology. This deregulation is demonstrated using HCHWA-D brain material. At the gene expression level, on individual genes, TGFβ1, its receptors and TGFβ-induced genes are upregulated (chapter 3). At the pathway level, in the transcriptomic study, extracellular matrix (ECM)-pathways related to TGFβ are increased (chapter 6). Lastly at the protein level using histology, we showed the actual activation of the TGFβ signaling cascade. First by showing the accumulation of its activated transcription factor (pSMAD2/3; chapter 3) in some angiopathic vessels, then by identifying the presence of the pro-fibrotic collagen 1 (col1) protein, a major TGFβ-induced genes, in amyloid-laden vessels (chapter 5). By selecting patients at different disease stages, we discovered that pSMAD2/3 and col1 accumulation in angiopathic vessel walls are correlated with CAA load (chapter 3 & 5).

We show that TGFβ deregulation correlates with disease severity. However, disease severity in HCHWA-D is also characterized by the higher occurrence of secondary remodelling of angiopathic vessels (1). In an initial study, we identified vascular calcifications, a specific secondary vascular remodelling, to be the cause of a recently described neuroimaging feature of advanced disease stage in HCHWA-D (2). In the subsequent study, we assessed the presence of osteopontin (OPN) in these calcifications, as described in other non-Aβ related vasculopathies in link with TGFβ increase (3). We further discovered a topographical association of pSMAD2/3 and OPN in these calcifications and we also linked their gradual accumulation with the calcification process (chapter 5).

Study of pathomechanisms in human brain tissue

material

Advantages & Difficulties

The use of human brain material is undeniably a unique chance to study pathomechanisms in relevant tissue and has a high scientific value. Using patient tissue gives access to the whole complexity of the disease in comparison to pathological processes in vitro which lack the cellular and physiological context or in vivo studies in animal model which cannot fully recapitulate human brain diseases.

Despite these advantages, patient brain material is underused in research mostly because its utilization remains challenging (4). The principal drawback is the heterogeneity of samples due to individual differences (age, gender, health, body mass, life style), prior medication exposure and additional co-morbidities, which are confounding factors. Secondly, for gene expression analysis, the RNA quality is variable, mostly due to the circumstances of death (agonal stress, (5)) but other factors such as postmortem interval, dissection protocols and storage conditions can influence the RNA integrity as well. Because the RNA quality is generally lower than for RNA isolated from cell cultures or animal models, it requires testing and optimisation of standard procedures, as discussed in chapter 6. Thirdly, the material is scarce, which might be restrictive in experimental design. Lastly, postmortem human tissue reflects the end-stage of the disease and therefore cannot replace cellular and animal research models where early events in illness can be studied.

Frontal versus occipital pathology in HCHWA-D

(4)

Discussion

The correlation of the different CAA load with pSMAD2/3 accumulation independently of the brain area, revealed TGFβ-related pathomechanisms (chapter 3).

• Pathological disease severity in HCHWA-D is comprised of CAA load plus additional secondary vascular remodeling

• Gene expression in frontal and occipital cortex is similar but secondary vessel pathology showed more perivascular changes and vascular calcifications occipitally

Consequences of TGFβ activation

TGFβs are pleiotropic cytokines involved in a variety of biological effects, modulating ECM metabolism, cell cycle as well as immune response (6). Transgenic mice overexpressing TGFβ1 in brain cells have been extensively studied to understand the influence of this cytokine on the vasculature, on neuroinflammation and on Aβ deposition (7,8). Results of this thesis, in light of knowledge from previous neuropathological and transgenic mice studies, further implicate TGFβ in HCHWA-D phenotypic variability.

Effects of TGFβ on the cerebrovasculature

Cerebrovascular fibrosis

The pathogenesis of cerebrovascular fibrosis has been studied in mouse model overexpressing TGFβ1 in astrocytes (GFAP-TGFβ1, (9)). It starts by a perivascular astrogliosis inducing an early thickening of vascular basement membranes (3-4 months) by ECM proteins, followed by the deposition of amyloid fibrils by 6 months of age. When these mice are crossed with mice overexpressing human APP (hAPP) in neurons (GFAP-TGFβ1/PDGF-hAPP), amyloid vascular deposits are detected even earlier (2-3 months). Remarkably, another mouse model with a neuron-specific TGFβ1 inducible overexpression, develop a similar angiopathic phenotype via induction of perivascular astrogliosis, confirming that astrocytes are major player in the cerebrovascular fibrosis (10).

Prior studies in HCHWA-D also described perivascular astrogliosis associated with arteriolar CAA (11) and perivascular ECM remodelling (12,13). Although the chronology of events is unknown, based on animal model studies, a TGFβ1-mediated fibrosis, preceding and triggering the CAA pathology might be an early event in HCHWA-D patients as well.

Accordingly, our transcriptomic study suggests an early TGFβ1-mediated fibrosis. By comparing the human transcriptome with the transcriptome of young APP-E693Q mice at the pathway level, we could identify an overlap

in the upregulation of ECM-related pathways, which is in mice prior to the onset of CAA pathology (chapter 6). Moreover, we confirmed in patient material the increase of TGFβ-induced pro-fibrotic target genes such as plasminogen activator inhibitor-1 (PAI-1), fibronectin (FN1) and collagen (Type I Col1A1 and Type III Col3A1; chapter 3). We further proposed that the accumulation of Col1 protein in the vessel walls might be prior to amyloid deposition (chapter 5), suggesting a TGFβ1-mediated fibrosis triggering the CAA pathology.

Secondary vascular remodelling

Prolonged TGFβ1 overexpression in astrocytic-TGFβ1 mice results in degenerative changes in microvascular cells (at 9-18 month) and cerebral microhemorrhages at older age (18-24 month). Our results show that TGFβ deregulation correlates with disease severity. As most severe cases are in general the oldest patients, it suggests that also in patients TGFβ activation is prolonged, and is likely modulating the CAA pathology. The most severe cases also present with secondary vascular remodeling such as vascular calcifications studied in this thesis. Interestingly, in the neuronal-TGFβ1 mice, a microarray analysis of cortical gene expression showed upregulation of OPN as well as various genes also involved in tissue mineralization and vascular calcification, strengthening the influence of TGFβ1 on this type of vasculopathy.

Effect of TGFβ on microglia / macrophages phenotype

TGFβ is also a major player in the inflammatory phenotype of brain microglia and macrophages. The inflammatory response can present either two or three states: a pro-inflammatory state (M1) and a repair/resolution state (M2), which is sometimes subdivided into an alternative inflammation state (phagocytic, fibrotic) or an acquired deactivation state (immunosuppressive) (14). The M2 state is associated with, and induced by, a TGFβ1 increase (15). Although not extensively studied in this thesis, the immune response in HCHWA-D is thought to be influenced by TGFβ1 upregulation and to modulate the phenotype.

(5)

141 140

Discussion

state. In HCHWA-D patients, typical M2 genes Sphk1, TGFβ1 and HO-1 (acquired deactivation) together with chitinase-Like Proteins genes CHI3L2 and CHI3L1 (alternative inflammatory, (14)) were significantly increased in our RNAseq study (chapter 6, supplementary tables).

Lastly, inflammatory CAA can also be induced by TGFβ pathway activation. Inflammatory CAA is characterized by the infiltration of macrophages and other blood-derived immune cells around and/or within the vessel walls of CAA-affected blood vessels. Perivascular macrophages have a function in the regulation of CAA (17) and TGFβ1 is a central player in the modulation of the phenotype and brain infiltration of macrophages. Indeed, in APP and TGFβ1-overexpressing mice, induction of inflammatory CAA via activation of macrophages reduces the CAA pathology and is thus considered as beneficial (18,19). At histopathological examination, inflammatory CAA is common in HCHWA-D and is more frequently detected in patients with the most severe phenotype (20). Whether inflammatory CAA is beneficial or detrimental to the patients is a matter of debate. Although it is associated with diverse vasculopathies and increased risk of dementia (16), patients with inflammatory CAA survive on average longer and have smaller non-fatal hemorrhages (M. Maat Schieman, unpublished work). In conclusion, the exact contribution of these immune cells to the CAA pathology is unclear, limited to animal models studies and requires further investigations.

• The TGFβ pathway is activated in HCHWA-D, and contributes to the phenotypic variability

• TGFβ chronic overexpression aggravates the vascular fibrosis, is associated with the CAA pathology severity and might promote vascular calcification

Possible causes of TGFβ activation in HCHWA-D

TGFβ in the central nervous system (CNS) is induced upon injuries as part of the healing process. Independently of stroke-related brain injuries, we discuss below potential early TGFβ induction in HCHWA-D triggered by cellular stress or interference with TGFβ related processes.

Early response to cellular toxicity

Oligomeric Aβ species (oAβs) have been extensively studied and are the most toxic of the Aβ species accumulating in AD and in APP-overexpressing animal models. oAβ are potent neurotoxins inducing a wide range of cellular

dysfunctions (oxidative and ER stress, calcium dyshomeostasis, cell cycle re-entry, mitochondrial dysfunction) as well as neurodegeneratives reactions (21,22).

A higher stability of oAβs due to the E22Q mutation (chapter 1) have been predicted in silico (23), demonstrated in vitro (24–26) and further found in

vivo, in APP-E693Q mice (27). In APP-E693∆ mice, a closely related animal

model with a lack of glutamate at position 22 of Aβ , intraneuronal oAβs lead to mitochondrial dysfunction (28). Even though the actual existence of oAβs has never been proven in patients, we also found a mitochondrial dysfunction in our HCHWA-D transcriptomic study (chapter 6) indicating cellular stress, possibly triggered by toxic oligomeric species.

While less investigated, oAβs can elicit a stress response in other cell types than neurons. Particularly, endothelial cells (ECs) in vitro are highly susceptible to Dutch oAβ peptide (29) and endothelial injuries lead to a CAA phenotype associated with TGFβ1 upregulation (30). Nevertheless Dutch oAβ addition to ECs in monoculture leads to a decrease in TGFβ1 (31,32), and it is likely that the TGFβ response observed in vivo mostly comes from other cells type such as glial cells (33). Especially reactive astrocytes are susceptible to oAβs toxic species and can induce a neuroprotective TGFβ response. A recent study revealed that oAβ can activate astrocytes; astrocyte-derived TGFβ1 prevents synapse loss induced by oAβ in AD mice (34). Such a protective astrocytic reaction to limit the level of neurotoxic soluble oAβ was also suggested to occur in HCHWA-D (11).

Direct interferences with TGFβ activity

Finally, the Dutch mutation could directly interfere with TGFβ-related processes.

TGFβ1 co-aggregates with amyloid parenchymal plaques in AD and Down Syndrome, suggesting a neuroprotective Aβ sequestration (35). It was demonstrated that Aβ can bind to TGFβ (36) and to TGFβ receptors (TGFβR) (37). Although the effect of the Dutch Aβ peptide on the interaction with TGFβ is unknown, the FAQD Dutch Aβ motif was predicted to influence the Aβ binding to TGFβ receptors, meaning a putative direct interference with the signalling pathway.

(6)

Discussion

APP23 and APP Dutch mice (40)). Since TGFβ1 is also stored in large amount in platelets and Col1 particularly can induce platelet exocytosis (41), the presence of vascular Col1 in HCHWA-D (chapter 5) might enhance platelet recruitment and exocytosis, thereby providing local higher TGFβ1 and APP concentrations and further aggravating (or even initiating) the CAA pathology.

• Pathomechanisms such as enhanced formation of oAβ, endothelial injuries or coagulation disorders could enhance endogenous TGFβ response before stroke onset

Future perpectives

Pathomechanisms in HCHWA-D

A major question raised by this work is whether the Dutch mutation itself triggers an endogenous TGFβ increase.

As described above, TGFβ1 cerebral overexpression in AD mice induces a CAA pathology increase and a parenchymal amyloid reduction. Intriguingly, a CAA phenotype is consistently induced in transgenic animals upon insertion of Dutch mutation in the APP sequence (APP Dutch (42), APP Dutch/ APP23 (43); APP Dutch/BACE1 (44); APP-E693Q/PS1∆exon9 (27); Knock-In model APP DSL (45)). TGFβ1 levels in those models would be interesting to measure in order to know whether the insertion of the Dutch mutation leads to an endogenous TGFβ1 increase as proposed in this thesis.

The direct effect of the Dutch Aβ peptide on TGFβ-related mechanisms could be further tested experimentally. As described in this chapter, some biological processes might only be triggered by specific Aβ species such as oAβs. Obtaining specific Aβ species in vitro using synthetic Aβ is problematic because Aβ peptide is highly unstable (personal observations). Instead, endogenous Aβ produced by iPS patient-derived cells, combined with recent progress in organoids or organ-on-chip technologies, opens new possibilities. Once established, an endogenous TGFβ increase, for example, in patient-derived astrocyte/neuronal cocultures would be an interesting phenotype for further studies into pathomechanisms or therapeutic targets. Biomarkers of cerebrovascular fibrosis

New biomarkers of CAA pathology in blood and cerebrospinal fluid (CSF) would be highly valuable to detect the disease onset, improve the clinical diagnosis and understand underlying pathomechanisms.

Results from this thesis suggest that biomarkers of cerebrovascular fibrosis such as direct measure of TGFβ levels or detection of downstream pro-fibrotic components (such as FN1, Serpine1 and OPN) could be of interest. Brain TGFβ levels in asymptomatic mutation carriers and in post-stroke patients have not been determined to date. In mutation carriers, detection of TGFβ levels might indicate the onset of brain injuries (see Graphic summary) and help in defining windows for therapeutic intervention. For clinical practice, biomarkers of cerebrovascular fibrosis could help to estimate disease severity and progression. Finally, monitoring such biomarkers in therapeutic intervention studies will add insight into safety, effect and effectiveness of drugs in clinical trials.

Developing biomarkers of cerebrovascular fibrosis would help to better estimate the onset and progression of CAA pathology in HCHWA-D but also in other CAA-related vasculopathy and small vessel diseases where TGFβ plays a central role as well (46,47).

• Developing biomarkers of cerebrovascular fibrosis will help understanding the disease in its initial phase, improving diagnostic tools and defining the best therapeutic strategies for HCHWA-D • Such biomarkers are of interest for other CAA pathologies

Therapeutics intervention

TGFβ lowering therapies

TGFβ pathway is a double-edged sword

TGF-β is key in tissue homeostasis and disruption of the TGFβ pathway has been implicated in numerous human diseases. Activation of the TGFβ pathway in systemic diseases (such as atherosclerosis) has beneficial anti-inflammatory properties as well as detrimental profibrotic effects. Similar dual effects have been observed in the CNS. In animal models, TGFβ1 increase is neuroprotective, especially acutely after injury (48–51). However, chronically high levels cause extensive cerebrovascular fibrosis as described in this thesis.

Considerations with TGFβ lowering therapies

(7)

145 144

Discussion

context of stroke, acute TGFβ upregulation restricts the infarct size in mouse models (49,55). However, in ageing animals, the TGFβ response is stronger and last longer (56,57) resulting in more long-term detrimental effects. Maintaining TGFβ levels within a certain range might therefore slow down CAA severity and disease progression. In practice, strategies targeting the downstream ECM modulators and pro-fibrotic genes would be a safer option than interfering with the TGFβ signaling. For example PSTG2 (COX-2) inhibitor celecoxib can lower FN1 and Col1 expression and attenuate the vessel wall thickness in stroke-prone spontaneously hypertensive rats (58).

Amyloid lowering therapy

Anti-Aβ immunotherapy

Two strategies of Aβ vaccination have been tried in AD clinical trial, either active immunization with Aβ species to trigger a host immune response, or passive immunization by direct infusion of anti-Aβ antibodies.

But although anti-Aβ immunotherapies are effective in removing parenchymal Aβ, an increase in CAA pathology is observed, implicating a TGFβ upregulation. Amyloid-related imaging abnormalities (or ARIA) in amyloid-modifying therapeutic trials are a major concern and multiple studies indicate that vascular amyloid is the common pathophysiological mechanism behind ARIA (59). At autopsy, Aβ vaccination consistently induced a removal of parenchymal plaques and an increase in amyloid vascular deposition in brains of clinical trial participants (60,61). Anti-Aβ immunotherapy in old AD transgenic mice had similar effects, with a worsening in CAA pathology and hemorrhages with longer treatment. Interestingly this phenotype is again triggered by a change in the inflammatory state characterized by TGFβ upregulation (62).

A first immunotherapy trial for probable CAA patients with Ponezumab, an antibody-targeting Aβ40, was launched by Pfizer in 2013 (NCT01821118; https://clinicaltrials.gov). After promising results in mouse models with CAA pathology (63), infusions of humanized ponezumab or placebo were evaluated for changes in occipital vascular reactivity by BOLD fMRI (primary endpoint). Although Ponezumab was well tolerated and showed drug-placebo differences, it did not meet the primary endpoint and the trial was stopped in 2016 in phase II (results not yet published; (64)). The pathological effect of this therapy is unknown with the current biomarkers and understanding of pathomechanisms. Based on the loss of cerebrovascular reactivity in the treated patients (65) one might speculate that the treatment resulted in a possible aggravation of the CAA pathology similarly to the AD clinical trial observations. In the light of this thesis and previous literature (66), the loss of cerebrovascular reactivity could be associated with an increase in TGFβ mediated vascular fibrosis.

Other anti-Aβ strategies

Dutch amyloid seeds are harmful and irreversible due to a prion-like effect on WT Aβ (67), therefore an early intervention is preferable. One strategy, recently described for AD (68) and currently developed in our group for HCHWA-D, uses APP exon skipping to reduce the total Aβ secretion. Targeting cerebral genes with exon skipping is feasible and already in clinical trial for Huntington’s Disease, where a recent clinical trial showed very promising results (69,70).

Alternative therapeutic strategies

Even though early anti-Aβ treatments are likely to be beneficial, therapeutic strategies uniquely focused on amyloid reduction will never completely prevent the CAA pathology in the context of vascular fibrosis. The complexity of CAA pathologies when amyloid deposition and cerebrovascular fibrosis are mixed is illustrated by studies in bitransgenic APP/TGFβ (A/T) mice (71). Therapies which were beneficial in single APP or TGFβ mice have a different outcome in A/T mice because vascular impairment caused by Aβ and TGFβ are mechanistically different (72). In HCHWA-D and particularly in sporadic CAA, where ageing plays an important role, combinatorial therapeutic treatments simultaneously targeting soluble toxic amyloid species (such as Taxifolin targeting oAβs (73)) and preventing the cerebrovascular remodeling would in all likelihood be more effective than single target treatments.

• TGFβ lowering therapies in brain should be considered with care • Treatment to reduce the cerebrovascular fibrosis would be a safer

option

(8)

Discussion

Graphic summary: hypothetical pathomechanisms linking brain TGFβ increase with

CAA progression. An early response to cellular toxicity could initiate the TGFβ increase (Hit 1) and trigger the vascular fibrosis in the pre-symptomatic phase. Acute stroke-related TGFβ increase (Hit 2) could further accelerate the disease progression; leading in final stage to calcification of some angiopathic vessels.

References

1. Vinters H V., Natté R, Maat-Schieman MLC, Van Duinen SG, Hegeman-Kleinn I, Welling-Graafland C, et al. Secondary microvascular degeneration in amyloid angiopathy of patients with hereditary cerebral hemorrhage with amyloidosis, Dutch type (HCHWA-D). Acta Neuropathol. 1998 Mar;95(3):235–44.

2. Koemans EA, van Etten ES, van Opstal AM, Labadie G, Terwindt GM, Wermer MJH, et al. Innovative Magnetic Resonance Imaging Markers of Hereditary Cerebral Amyloid Angiopathy at 7 Tesla. Stroke. 2018 Apr; 49(6):1518–20.

3. Mohler ER, Adam LP, McClelland P, Graham L, Hathaway DR. Detection of osteopontin in calcified human aortic valves. Arterioscler Thromb Vasc Biol. 1997 Mar;17(3):547–52. 4. Mccullumsmith RE, Hammond JH, Shan D, Meador-Woodruff JH. Postmortem brain: An underutilized substrate for studying severe mental illness. Neuropsychopharmacology. 2014 Jan;39(1):65–87.

5. Tomita H, Vawter MP, Walsh DM, Evans SJ, Choudary P V., Li J, et al. Effect of agonal and postmortem factors on gene expression profile: Quality control in microarray analyses of postmortem human brain. Biol Psychiatry. 2004 Feb 15;55(4):346–52.

6. Vivien D, Ali C. Transforming growth factor-β signalling in brain disorders. Cytokine Growth Factor Rev. 2006;17(1–2):121–8.

7. Buckwalter M, Pepper JP, Gaertner RF, Von Euw D, Lacombe P, Wyss-Coray T. Molecular and functional dissection of TGF-β1-induced cerebrovascular abnormalities in transgenic mice. In: Annals of the New York Academy of Sciences. 2002. p. 87–95.

8. Buckwalter MS, Wyss-Coray T. Modelling neuroinflammatory phenotypes in vivo. J Neuroinflammation. 2004 Jul 1;1(1):10.

9. Wyss-Coray T, Lin C, Sanan DA, Mucke L, Masliah E. Chronic overproduction of transforming growth factor-β1 by astrocytes promotes Alzheimer’s disease-like microvascular degeneration in transgenic mice. Am J Pathol. 2000 Jan;156(1):139–50. 10. Ueberham U, Ueberham E, Brückner MK, Seeger G, Gärtner U, Gruschka H, et al.

Inducible neuronal expression of transgenic TGF-β1 in vivo: Dissection of short-term and long-term effects. Eur J Neurosci. 2005 Jul;22(1):50–64.

11. Maat-Schieman MLC, Yamaguchi H, Hegeman-Kleinn IM, Welling-Graafland C, Natté R, Roos RAC, et al. Glial reactions and the clearance of amyloid β protein in the brains of patients with hereditary cerebral hemorrhage with amyloidosis-Dutch type. Acta Neuropathol. 2004 May;107(5):389–98.

12. van Duinen SG, Maat-Schieman MLC, Bruijn JA, Haan J, Roos RAC, Wattendorff AR, et al. Cortical Tissue of Patients With Hereditary Cerebral-Hemorrhage With Amyloidosis (Dutch) Contains Various Extracellular-Matrix Deposits. Lab Investig. 1995 Aug;73(2):183–9.

13. Wilhelmus MMM, Bol JGJM, van Duinen SG, Drukarch B. Extracellular matrix modulator lysyl oxidase colocalizes with amyloid-beta pathology in Alzheimer’s disease and hereditary cerebral hemorrhage with amyloidosis-Dutch type. Exp Gerontol. 2013 Feb;48(2):109–14.

(9)

149 148

Discussion

15. Heneka MT, Carson MJ, Khoury J El, Landreth GE, Brosseron F, Feinstein DL, et al. Neuroinflammation in Alzheimer’s disease. Lancet Neurol. 2015;14(4):388–405. 16. Natté R, Maat-Schieman MLC, Haan J, Bornebroek M, Roos RAC, Van Duinen SG.

Dementia in hereditary cerebral hemorrhage with amyloidosis-Dutch type is associated with cerebral amyloid angiopathy but is independent of plaques and neurofibrillary tangles. Ann Neurol. 2001 Dec;50(6):765–72.

17. Hawkes CA, McLaurin J. Selective targeting of perivascular macrophages for clearance of -amyloid in cerebral amyloid angiopathy. Proc Natl Acad Sci. 2009;106(4):1261–6. 18. Town T, Laouar Y, Pittenger C, Mori T, Szekely CA, Tan J, et al. Blocking TGF-β–Smad2/3

innate immune signaling mitigates Alzheimer-like pathology. Nat Med. 2008;14(6):681–7. 19. Lifshitz V, Weiss R, Benromano T, Kfir E, Blumenfeld-Katzir T, Tempel-Brami C, et al. Immunotherapy of cerebrovascular amyloidosis in a transgenic mouse model. Neurobiol Aging. 2012 Feb;33(2):432.e1-432.e13.

20. Vinters H V. Inflammation complicates an “age-related” cerebral microangiopathy. Can J Neurol Sci. 2011 Jul;38(4):543–4.

21. Viola KL, Klein WL. Amyloid β oligomers in Alzheimer’s disease pathogenesis, treatment, and diagnosis. Acta Neuropathol. 2015;129(2):183–206.

22. DiChiara T, DiNunno N, Clark J, Bu R Lo, Cline EN, Rollins MG, et al. Alzheimer’s Toxic Amyloid Beta Oligomers: Unwelcome Visitors to the Na/K ATPase alpha3 Docking Station. Yale J Biol Med. 2017 Mar;90(1):45–61.

23. Kassler K, Horn AHC, Sticht H. Effect of pathogenic mutations on the structure and dynamics of Alzheimer’s A beta 42-amyloid oligomers. J Mol Model. 2010 May;16(5):1011–20. 24. Miravalle L, Tokuda T, Chiarle R, Giaccone G, Bugiani O, Tagliavini F, et al. Substitutions

at codon 22 of Alzheimer’s abeta peptide induce diverse conformational changes and apoptotic effects in human cerebral endothelial cells. J Biol Chem. 2000 Sep 1;275(35):27110–6.

25. Ohshima Y, Taguchi K, Mizuta I, Tanaka M, Tomiyama T, Kametani F, et al. Mutations in the β-amyloid precursor protein in familial Alzheimer’s disease increase Aβ oligomer production in cellular models. Heliyon. 2018;(October 2017):e00511.

26. Sian a K, Frears ER, El-Agnaf OM, Patel BP, Manca MF, Siligardi G, et al. Oligomerization of beta-amyloid of the Alzheimer’s and the Dutch-cerebral-haemorrhage types. Biochem J. 2000;349:299–308.

27. Gandy S, Simon AJ, Steele JW, Lublin AL, Lah JJ, Walker LC, et al. Days to criterion as an indicator of toxicity associated with human Alzheimer amyloid-beta oligomers. Ann Neurol. 2010 Aug;68(2):220–30.

28. Umeda T, Tomiyama T, Sakama N, Tanaka S, Lambert MP, Klein WL, et al. Intraneuronal amyloid beta oligomers cause cell death via endoplasmic reticulum stress, endosomal/ lysosomal leakage, and mitochondrial dysfunction in vivo. J Neurosci Res. 2011 Jul;89(7):1031–42.

29. Viana RJS, Nunes a F, Castro RE, Ramalho RM, Meyerson J, Fossati S, et al. Tauroursodeoxycholic acid prevents E22Q Alzheimer’s Aβ toxicity in human cerebral endothelial cells. Cell Mol Life Sci. 2009;66(6):1094–104.

30. Tan X-L, Xue Y-Q, Ma T, Wang X, Li JJ, Lan L, et al. Partial eNOS deficiency causes spontaneous thrombotic cerebral infarction, amyloid angiopathy and cognitive impairment. Mol Neurodegener. 2015;10(1):24.

31. Paris D, Ait-Ghezala G, Mathura VS, Patel N, Quadros A, Laporte V, et al. Anti-angiogenic activity of the mutant Dutch A(beta) peptide on human brain microvascular endothelial cells. Brain Res Mol Brain Res. 2005 May 20;136(1–2):212–30.

32. Solito R, Corti F, Fossati S, Mezhericher E, Donnini S, Ghiso J, et al. Dutch and Arctic mutant peptides of beta amyloid(1-40) differentially affect the FGF-2 pathway in brain endothelium. Exp Cell Res. 2009 Mar 1;315(3):385–95.

33. Liddelow SA, Guttenplan KA, Clarke LE, Bennett FC, Bohlen CJ, Schirmer L, et al. Neurotoxic reactive astrocytes are induced by activated microglia. Nature. 2017;541(7638):481–7.

34. Diniz LP, Tortelli V, Matias I, Morgado J, Bergamo Araujo AP, Melo HM, et al. Astrocyte Transforming Growth Factor Beta 1 Protects Synapses against Abeta Oligomers in Alzheimer’s Disease Model. J Neurosci. 2017 Jul;37(28):6797–809.

35. van der Wal EA, Gomez-Pinilla F, Cotman CW. Transforming growth factor-beta 1 is in plaques in Alzheimer and Down pathologies. Neuroreport. 1993 Jan;4(1):69–72. 36. Mousseau DD, Chapelsky S, De Crescenzo G, Kirkitadze MD, Magoon J, Inoue S, et al.

A direct interaction between transforming growth factor (TGF)-betas and amyloid-beta protein affects fibrillogenesis in a TGF-beta receptor-independent manner. J Biol Chem. 2003 Oct 3;278(40):38715–22.

37. Huang SS, Huang FW, Xu J, Chen S, Hsu CY. Amyloid beta-Peptide Possesses a Transforming Growth Factor-beta Activity *. 1998;273(42):27640–4.

38. Bornebroek M, Von Dem Borne PAK, Haan J, Meijers JCM, Van Nostrand WE, Roos RAC. Binding of amyloid β precursor protein to coagulation factor XIa in vivo may favour haemorrhagic stroke. J Neurol. 1998 Feb;245(2):111–5.

39. Rozemuller AJ, Roos RA, Bots GT, Kamphorst W, Eikelenboom P, Van Nostrand WE. Distribution of beta/A4 protein and amyloid precursor protein in hereditary cerebral hemorrhage with amyloidosis-Dutch type and Alzheimer’s disease. Am J Pathol. 1993;142(5):1449–57.

40. Gowert NS, Donner L, Chatterjee M, Eisele YS, Towhid ST, Münzer P, et al. Blood platelets in the progression of alzheimer’s disease. PLoS One. 2014;9(2).

41. Alberio L, Dale GL. Flow cytometric analysis of platelet activation by different collagen types present in the vessel wall. Br J Haematol. 1998 Sep;102(5):1212–8.

42. Herzig MC, Winkler DT, Burgermeister P, Pfeifer M, Kohler E, Schmidt SD, et al. Aβ is targeted to the vasculature in a mouse model of hereditary cerebral hemorrhage with amyloidosis. Nat Neurosci. 2004 Oct;7(9):954–60.

43. Herzig MC, Eisele YS, Staufenbiel M, Jucker M. E22Q-mutant Aβ peptide (AβDutch) increases vascular but reduces parenchymal Aβ deposition. Am J Pathol. 2009 Mar;174(3):722–6.

44. Herzig MC, Paganetti P, Staufenbiel M, Jucker M. BACE1 and mutated presenilin-1 differently modulate Aβ40 and Aβ42 levels and cerebral amyloidosis in APPDutch transgenic mice. Neurodegener Dis. 2007 Jan;4(2–3):127–35.

45. Li H, Guo Q, Inoue T, Polito V a, Tabuchi K, Hammer RE, et al. Vascular and parenchymal amyloid pathology in an Alzheimer disease knock-in mouse model: interplay with cerebral blood flow. Mol Neurodegener. 2014;9:28.

(10)

Discussion

47. Müller K, Courtois G, Ursini MV, Schwaninger M. New Insight Into the Pathogenesis of Cerebral Small-Vessel Diseases. Stroke. 2017;STROKEAHA.116.012888.

48. Flanders KC, Ren RF, Lippa CF. Transforming growth factor-betas in neurodegenerative disease. Prog Neurobiol. 1998;54(1):71–85.

49. Doyle KP, Cekanaviciute E, Mamer LE, Buckwalter MS. TGFβ signaling in the brain increases with aging and signals to astrocytes and innate immune cells in the weeks after stroke. J Neuroinflammation. 2010 Jan;7(1):62.

50. Cekanaviciute E, Fathali N, Doyle KP, Williams AM, Han J, Buckwalter MS. Astrocytic transforming growth factor-beta signaling reduces subacute neuroinflammation after stroke in mice. Glia. 2014;62(8):1227–40.

51. Dobolyi A, Vincze C, Pál G, Lovas G. The neuroprotective functions of transforming growth factor Beta proteins. Int J Mol Sci. 2012 Jan;13(7):8219–58.

52. Zhang ZY, Li C, Zug C, Schluesener HJ. Icariin ameliorates neuropathological changes, TGF-β1 accumulation and behavioral deficits in a mouse model of cerebral amyloidosis. PLoS One. 2014;9(8).

53. Sachdeva AK, Chopra K. Lycopene abrogates Abeta (1-42)-mediated neuroinflammatory cascade in an experimental model of Alzheimer’s disease. J Nutr Biochem. 2015;26(7):736–44. 54. Li C, Zug C, Qu H, Schluesener H, Zhang Z. Hesperidin ameliorates behavioral impairments

and neuropathology of transgenic APP/PS1 mice. Behav Brain Res. 2015;281:32–42. 55. Song L, Liu F, Liu C, Li X, Zheng S, Li Q, et al. Neuroprotective effects of SMADs in a

rat model of cerebral ischemia/reperfusion. Neural Regen Res. 2015;10(3):438.

56. Lively S, Schlichter LC. Age-Related Comparisons of Evolution of the Inflammatory Response After Intracerebral Hemorrhage in Rats. Transl Stroke Res. 2012;3(SUPPL. 1):132–46.

57. Buga AM, Margaritescu C, Scholz CJ, Radu E, Zelenak C, Popa-Wagner A. Transcriptomics of post-stroke angiogenesis in the aged brain. Front Aging Neurosci. 2014;6(MAR):1–20. 58. Tang J, Xiao W, Li Q, Deng Q, Chu X, Chen Y, et al. A Cyclooxygenase-2 Inhibitor

Reduces Vascular Wall Thickness and Ameliorates Cognitive Impairment in a Cerebral Small Vessel Diseases Rat Model. Vol. 12, Current Alzheimer Research. 2015. p. 704–10. 59. Sperling RA, Jack CRJ, Black SE, Frosch MP, Greenberg SM, Hyman BT, et al. Amyloid-related imaging abnormalities in amyloid-modifying therapeutic trials: recommendations from the Alzheimer’s Association Research Roundtable Workgroup. Alzheimers Dement. 2011 Jul;7(4):367–85.

60. Zotova E, Bharambe V, Cheaveau M, Morgan W, Holmes C, Harris S, et al. Inflammatory components in human Alzheimer’s disease and after active amyloid- 42 immunization. Brain. 2013 Aug 13;

61. Boche D, Zotova E, Weller RO, Love S, Neal JW, Pickering RM, et al. Consequence of Abeta immunization on the vasculature of human Alzheimer’s disease brain. Brain. 2008 Dec;131(Pt 12):3299–310.

62. Wilcock DM, Rojiani A, Rosenthal A, Subbarao S, Freeman MJ, Gordon MN, et al. Passive immunotherapy against Aβ in aged APP-transgenic mice reverses cognitive deficits and depletes parenchymal amyloid deposits in spite of increased vascular amyloid and microhemorrhage. Vol. 1, Journal of Neuroinflammation. London; 2004. p. 24.

63. Bales KR, O’Neill SM, Pozdnyakov N, Pan F, Caouette D, Pi Y, et al. Passive immunotherapy targeting amyloid-β reduces cerebral amyloid angiopathy and improves vascular reactivity. Brain. 2016;139(2):563–77.

64. Banerjee G, Carare R, Cordonnier C, Greenberg SM, Schneider JA, Smith EE, et al. The increasing impact of cerebral amyloid angiopathy: Essential new insights for clinical practice. J Neurol Neurosurg Psychiatry. 2017;88(11):982–94.

65. Leurent C, Goodman JA, Zhang Y, He P, Polimeni JR, Gurol ME, et al. Immunotherapy with ponezumab for probable cerebral amyloid angiopathy. Ann Clin Transl Neurol. 2019;6(4):795–806.

66. Tong X-K. Vascular Remodeling versus Amyloid -Induced Oxidative Stress in the Cerebrovascular Dysfunctions Associated with Alzheimer’s Disease. J Neurosci. 2005 Nov 30;25(48):11165–74.

67. Condello C, Lemmin T, Stöhr J, Nick M, Wu Y, Maxwell AM, et al. Structural heterogeneity and intersubject variability of Aβ in familial and sporadic Alzheimer’s disease. Proc Natl Acad Sci. 2018;201714966.

68. Chang JL, Hinrich AJ, Roman B, Norrbom M, Rigo F, Marr RA, et al. Targeting Amyloid-beta Precursor Protein, APP, Splicing with Antisense Oligonucleotides Reduces Toxic Amyloid-beta Production. Mol Ther. 2018 Jun;26(6):1539–51.

69. Rutten JW, Dauwerse HG, Peters DJM, Goldfarb A, Venselaar H, Haffner C, et al. Therapeutic NOTCH3 cysteine correction in CADASIL using exon skipping: In vitro proof of concept. Brain. 2016 Apr;139(4):1123–35.

70. Toonen LJA, Rigo F, van Attikum H, van Roon-Mom WMC. Antisense Oligonucleotide-Mediated Removal of the Polyglutamine Repeat in Spinocerebellar Ataxia Type 3 Mice. Mol Ther Nucleic Acids. 2017 Sep;8:232–42.

71. Ongali B, Nicolakakis N, Lecrux C, Aboulkassim T, Rosa-Neto P, Papadopoulos P, et al. Transgenic mice overexpressing APP and transforming growth factor-β1 feature cognitive and vascular hallmarks of Alzheimer’s disease. Am J Pathol. 2010 Dec;177(6):3071–80. 72. Hamel E. Cerebral Circulation: Function and Dysfunction in Alzheimer’s Disease.

J Cardiovasc Pharmacol. 2015;65(4):317–24.

Referenties

GERELATEERDE DOCUMENTEN

Chapter 5: Osteopontin and phospho-SMAD2/3 are associated with calcification of vessels in D-CAA, an hereditary cerebral amyloid angiopathy. Chapter 6: Brain Transcriptomic

Heparan sulfate proteoglycan expression in cerebrovascular amyloid deposits in Alzheimer’s disease and hereditary cerebral hemorrhage with amyloidosis (Dutch) brains.. Cortical

In January 2013, she started a PhD in Neurosciences with a focus on Hereditary cerebral hemorrhage with amyloidosis–Dutch type (HCHWA-D) under the combined guidance of

In the present study we aimed to investigate the potential of diffusion tensor magnetic reso- nance imaging (MRI) to determine alterations in the white matter of presymptomatic

In both HCHWA-D cases and the sCAA case, co-localization of Aβ with non-heme iron and calcium in the vessel wall was found in areas containing the striped cortex (Fig..

In this study, we aimed to confirm whether increased CSO-PVS may have an independent diagnostic role for CAA. For this purpose, we evaluated MRI-visible CSO-PVS in: 1) a

The relative gene expression levels of TGFb1 and TGFBR2 were significantly higher in the frontal and occipital lobes of HCHWA-D patients compared to age-related controls (Figure

Background and Purpose—The aim of the present study is to explore whether using 7 Tesla MRI, additional brain changes can be observed in Hereditary Cerebral Hemorrhage with